1
|
Tchio C, Williams J, Taylor H, Ollila H, Saxena R. An integrative approach prioritizes the orphan GPR61 genomic region in tissue-specific regulation of chronotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624721. [PMID: 39651283 PMCID: PMC11623522 DOI: 10.1101/2024.11.22.624721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Objectives Chronotype, a manifestation of circadian rhythms, affects morning or evening preferences and ease of getting-up. This study explores the genetic basis of morning chronotype and ease of getting-up, focusing on the G protein-coupled receptor locus, GPR61. Methods We analyzed the genetic correlation between chronotype and ease of getting-up using linkage disequilibrium score regression with summary statistics from the UK Biobank (n=453,379). We prioritized shared signals between chronotype and ease of getting-up using the Human Genetic Evidence (HuGE) score. We assessed the significance of GPR61 and the lead variant rs12044778 through colocalization and in-silico analyses from ENCODE, Genotype-Tissue Expression, Hi-C, and Knockout Mouse Project databases to explore potential regulatory roles of causal genes. Results We identified a strong genetic correlation (Rg=0.80, P=4.9 x10 324 ) between chronotype and ease of getting-up. Twenty-three genes, including three circadian core clock components, had high HuGE scores for both traits. Lead variant rs12044778 in GPR61 was prioritized for its high HuGE score (45) and causal pleiotropy (posterior probability=0.98). This morningness variant influenced gene expression in key tissues: decreasing GPR61 in tibial nerve, increasing AMIGO1 in subcutaneous adipose, and increasing ATXN7L2 in the cerebellum. Functional knockout models showed GPR61 knockout increased fat mass and activity, AMIGO1 knockout increased activity, and ATXN7L2 knockout reduced body weight without affecting activity. Conclusions Our findings reveal pleiotropic genetic factors influencing chronotype and ease of getting-up, emphasizing GPR61 's rs12044778 and nearby genes like AMIGO1 and ATXN7L2 . These insights advance understanding of circadian preferences and suggest potential therapeutic interventions. SIGNIFICANCE This study investigates the genetic underpinnings of chronotype preferences and ease of getting up, with a focus on the orphan G protein-coupled receptor GPR61 and the locus lead variant rs12044778. By combining genomic data with in silico functional analysis, we provide mechanistic insight into a locus for morning chronotype and ease of getting in the morning. We identified the variant rs12044778 as a key regulator of GPR61 and nearby genes AMIGO1 and ATXN7L2 influencing circadian and metabolic traits. Our findings shed light on the intricate genetic networks governing circadian rhythms, suggesting potential therapeutic targets for disorders of the circadian rhythm.
Collapse
|
2
|
Lees JA, Dias JM, Rajamohan F, Fortin JP, O'Connor R, Kong JX, Hughes EAG, Fisher EL, Tuttle JB, Lovett G, Kormos BL, Unwalla RJ, Zhang L, Dechert Schmitt AM, Zhou D, Moran M, Stevens KA, Fennell KF, Varghese AE, Maxwell A, Cote EE, Zhang Y, Han S. An inverse agonist of orphan receptor GPR61 acts by a G protein-competitive allosteric mechanism. Nat Commun 2023; 14:5938. [PMID: 37741852 PMCID: PMC10517971 DOI: 10.1038/s41467-023-41646-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 09/11/2023] [Indexed: 09/25/2023] Open
Abstract
GPR61 is an orphan GPCR related to biogenic amine receptors. Its association with phenotypes relating to appetite makes it of interest as a druggable target to treat disorders of metabolism and body weight, such as obesity and cachexia. To date, the lack of structural information or a known biological ligand or tool compound has hindered comprehensive efforts to study GPR61 structure and function. Here, we report a structural characterization of GPR61, in both its active-like complex with heterotrimeric G protein and in its inactive state. Moreover, we report the discovery of a potent and selective small-molecule inverse agonist against GPR61 and structural elucidation of its allosteric binding site and mode of action. These findings offer mechanistic insights into an orphan GPCR while providing both a structural framework and tool compound to support further studies of GPR61 function and modulation.
Collapse
Affiliation(s)
- Joshua A Lees
- Discovery Sciences, Medicine Design, Pfizer Inc., Groton, CT, USA
| | - João M Dias
- Discovery Sciences, Medicine Design, Pfizer Inc., Groton, CT, USA
| | | | | | - Rebecca O'Connor
- Discovery Sciences, Medicine Design, Pfizer Inc., Groton, CT, USA
| | - Jimmy X Kong
- Internal Medicine Research Unit, Pfizer Inc., Cambridge, MA, USA
| | - Emily A G Hughes
- Internal Medicine Research Unit, Pfizer Inc., Cambridge, MA, USA
| | - Ethan L Fisher
- Internal Medicine, Medicine Design, Pfizer Inc., Groton, CT, USA
| | - Jamison B Tuttle
- Internal Medicine, Medicine Design, Pfizer Inc., Cambridge, MA, USA
| | - Gabrielle Lovett
- Internal Medicine, Medicine Design, Pfizer Inc., Cambridge, MA, USA
| | - Bethany L Kormos
- Internal Medicine, Medicine Design, Pfizer Inc., Cambridge, MA, USA
| | | | - Lei Zhang
- Internal Medicine, Medicine Design, Pfizer Inc., Cambridge, MA, USA
| | | | - Dahui Zhou
- Internal Medicine, Medicine Design, Pfizer Inc., Groton, CT, USA
| | - Michael Moran
- Internal Medicine, Medicine Design, Pfizer Inc., Groton, CT, USA
| | | | | | | | - Andrew Maxwell
- Discovery Sciences, Medicine Design, Pfizer Inc., Groton, CT, USA
| | - Emmaline E Cote
- Discovery Sciences, Medicine Design, Pfizer Inc., Groton, CT, USA
| | - Yuan Zhang
- Internal Medicine, Medicine Design, Pfizer Inc., Cambridge, MA, USA
| | - Seungil Han
- Discovery Sciences, Medicine Design, Pfizer Inc., Groton, CT, USA.
| |
Collapse
|
3
|
Saito R, Bando T, Kotaniguchi M, Tamura T, Kuno T, Watanabe K, Mizukami Y, Kitamura S, Kadokawa H. Ethanolamine plasmalogens derived from whale brain stimulate both follicle-stimulating hormone and luteinizing hormone secretion by bovine gonadotrophs. Anim Sci J 2023; 94:e13839. [PMID: 37247943 DOI: 10.1111/asj.13839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/10/2023] [Accepted: 04/28/2023] [Indexed: 05/31/2023]
Abstract
Ethanolamine plasmalogens (EPls) are the only known ligands of a novel receptor, G protein-coupled receptor 61, and bovine brain EPls stimulate follicle-stimulating hormone (FSH) but not luteinizing hormone (LH), secreted by bovine gonadotrophs. We hypothesized that the brain EPls of whales (Balaenoptera edeni), another Cetartiodactyla with at least twice the lifespan of bovines, could stimulate FSH secretion by gonadotrophs. To test this hypothesis, bovine gonadotrophs (from approximately 2-year-old Japanese Black heifers) were cultured for 3.5 days and treated with increasing concentrations of brain EP1s from whales (approximately 22 years old). FSH and LH secretion was stimulated by all tested concentrations of whale EPls (p < 0.05). To clarify the important differences between bovine and whale EPls, we utilized two-dimensional liquid chromatography-mass spectrometry, which revealed 35 peaks. Among them, we observed significant differences between 12 EPl molecular species. Additionally, we identified differentially expressed genes for enzymes involved in EPl synthesis or degradation in the hypothalamus of young heifers and old cows (approximately 10 years old) as compared to whales (approximately 28 years old) via deep sequencing of the transcriptome. We conclude that whale brains contain unique EPls that stimulate both FSH and LH secretion by bovine gonadotrophs.
Collapse
Affiliation(s)
- Risa Saito
- Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | | | - Miyako Kotaniguchi
- International Polysaccharide Engineering Inc., Laboratory of Advanced Food Process Engineering, Organization for Research Promotion, Osaka Metropolitan University, Sakai, Japan
| | | | - Tomoe Kuno
- Institute of Cetacean Research, Tokyo, Japan
| | - Kenji Watanabe
- Center for Gene Research, Yamaguchi University, Ube, Japan
| | | | - Shinichi Kitamura
- Laboratory of Advanced Food Process Engineering, Organization for Research Promotion, Osaka Metropolitan University, Sakai, Japan
| | - Hiroya Kadokawa
- Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
4
|
Ethanolamine plasmalogens derived from scallops stimulate both follicle-stimulating hormone and luteinizing hormone secretion by bovine gonadotrophs. Sci Rep 2022; 12:16789. [PMID: 36202862 PMCID: PMC9537335 DOI: 10.1038/s41598-022-20794-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/19/2022] [Indexed: 11/08/2022] Open
Abstract
Brain ethanolamine plasmalogens (EPls) are the only known ligands of G-protein-coupled receptor 61, a novel receptor that stimulates follicle-stimulating hormone (FSH), but not luteinizing hormone (LH), secretion by bovine gonadotrophs. We hypothesized that the recently developed neuroprotective EPls extracted from scallop (Pecten yessoensis) (scallop EPls) could stimulate FSH secretion by gonadotrophs. To test this hypothesis, bovine gonadotrophs were cultured for 3.5 days and treated with increasing concentrations of scallop EPls. FSH secretion was stimulated by all tested concentrations of scallop EPls (P < 0.05). Surprisingly, LH secretion was stimulated by both 0.5 (P < 0.05) and 5 (P < 0.01) ng/mL of scallop EPls. To clarify the important differences between bovine brain and scallop EPls, we utilized two-dimensional liquid chromatography–mass spectrometry, which revealed 44 peaks, including 10 large peaks. Among them, eight were scallop-specific EPl molecular species, occupying approximately 58% of the total area percentage of scallop EPls. Almost all large peaks contained 4, 5, or 6 unsaturated double bonds in the carbon chain at the sn-2 position of the glycerol backbone. Our results showed that EPls from scallops, lacking pituitary glands, stimulated both FSH and LH secretion by bovine gonadotrophs.
Collapse
|
5
|
Chemosynthetic ethanolamine plasmalogen stimulates gonadotropin secretion from bovine gonadotrophs by acting as a potential GPR61 agonist. Anim Reprod Sci 2022; 241:106992. [DOI: 10.1016/j.anireprosci.2022.106992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 05/04/2022] [Accepted: 05/08/2022] [Indexed: 01/12/2023]
|
6
|
Feng F, Huang L, Zhou G, Wang J, Zhang R, Li Z, Zhang Y, Ba Y. GPR61 methylation in cord blood: a potential target of prenatal exposure to air pollutants. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2022; 32:463-472. [PMID: 32478566 DOI: 10.1080/09603123.2020.1773414] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 05/19/2020] [Indexed: 06/11/2023]
Abstract
To explore the impact of air pollutants exposure during pregnancy on infant DNA methylation, we identified correlated methylated genes in maternal and cord blood samples using the Illumina Human Methylation 27 k BeadChip. Quantitative methylation-specific PCR (QMS-PCR) was performed to validate the target gene methylation pattern in 568 participants. Then the association between air pollutants exposure and DNA methylation level in the target gene was investigated. The GPR61 gene with a higher methylation level both in mothers and newborns was identified as the target gene, and we found a positive mother-infant DNA methylation correlation in the promoter region of GPR61. Air pollutants exposure during entire pregnancy was associated with maternal and infant GPR61 DNA methylation. After adjusting confounding variables, maternal air pollutants exposure was still associated with infant GPR61 DNA methylation. In summary, GPR61 methylation in cord blood may be a potential target of prenatal exposure to air pollutants.
Collapse
Affiliation(s)
- Feifei Feng
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, China
| | - Li Huang
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, China
| | - Guoyu Zhou
- Department of Environmental Health, Zhengzhou University School of Public Health, Zhengzhou, China
| | - Jia Wang
- Department of Environmental Health, Zhengzhou University School of Public Health, Zhengzhou, China
| | - Ruiqin Zhang
- College of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou, China
| | - Zhiyuan Li
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, China
| | - Yawei Zhang
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- Department of Environmental Health Science, Yale School of Public Health, New Haven, CT, USA
| | - Yue Ba
- Department of Environmental Health, Zhengzhou University School of Public Health, Zhengzhou, China
| |
Collapse
|
7
|
Schachtschneider KM, Schook LB, Meudt JJ, Shanmuganayagam D, Zoller JA, Haghani A, Li CZ, Zhang J, Yang A, Raj K, Horvath S. Epigenetic clock and DNA methylation analysis of porcine models of aging and obesity. GeroScience 2021; 43:2467-2483. [PMID: 34523051 PMCID: PMC8599541 DOI: 10.1007/s11357-021-00439-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/09/2021] [Indexed: 12/29/2022] Open
Abstract
DNA-methylation profiles have been used successfully to develop highly accurate biomarkers of age, epigenetic clocks, for many species. Using a custom methylation array, we generated DNA methylation data from n = 238 porcine tissues including blood, bladder, frontal cortex, kidney, liver, and lung, from domestic pigs (Sus scrofa domesticus) and minipigs (Wisconsin Miniature Swine™). Samples used in this study originated from Large White X Landrace crossbred pigs, Large White X Minnesota minipig crossbred pigs, and Wisconsin Miniature Swine™. We present 4 epigenetic clocks for pigs that are distinguished by their compatibility with tissue type (pan-tissue and blood clock) and species (pig and human). Two dual-species human-pig pan-tissue clocks accurately measure chronological age and relative age, respectively. We also characterized CpGs that differ between minipigs and domestic pigs. Strikingly, several genes implicated by our epigenetic studies of minipig status overlap with genes (ADCY3, TFAP2B, SKOR1, and GPR61) implicated by genetic studies of body mass index in humans. In addition, CpGs with different levels of methylation between the two pig breeds were identified proximal to genes involved in blood LDL levels and cholesterol synthesis, of particular interest given the minipig's increased susceptibility to cardiovascular disease compared to domestic pigs. Thus, breed-specific differences of domestic and minipigs may potentially help to identify biological mechanisms underlying weight gain and aging-associated diseases. Our porcine clocks are expected to be useful for elucidating the role of epigenetics in aging and obesity, and the testing of anti-aging interventions.
Collapse
Affiliation(s)
- Kyle M. Schachtschneider
- Department of Radiology, University of Illinois At Chicago, Chicago, IL USA
- Department of Biochemistry and Molecular Genetics, University of Illinois At Chicago, Chicago, IL USA
- National Center for Supercomputing Applications, University of Illinois At Urbana-Champaign, Urban, IL USA
| | - Lawrence B. Schook
- Department of Radiology, University of Illinois At Chicago, Chicago, IL USA
- Department of Animal Sciences, University of Illinois At Urbana-Champaign, Urbana, IL USA
| | - Jennifer J. Meudt
- Biomedical & Genomic Research Group, Department of Animal and Dairy Sciences, University of Wisconsin – Madison, Madison, WI USA
| | - Dhanansayan Shanmuganayagam
- Biomedical & Genomic Research Group, Department of Animal and Dairy Sciences, University of Wisconsin – Madison, Madison, WI USA
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
| | - Joseph A. Zoller
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA USA
| | - Amin Haghani
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA USA
| | - Caesar Z. Li
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA USA
| | - Joshua Zhang
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Gonda Building, 695 Charles Young Drive South, Los Angeles, CA 90095 USA
| | - Andrew Yang
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA USA
| | - Ken Raj
- Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, UK
| | - Steve Horvath
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Gonda Building, 695 Charles Young Drive South, Los Angeles, CA 90095 USA
| |
Collapse
|
8
|
Powell DR, Revelli JP, Doree DD, DaCosta CM, Desai U, Shadoan MK, Rodriguez L, Mullens M, Yang QM, Ding ZM, Kirkpatrick LL, Vogel P, Zambrowicz B, Sands AT, Platt KA, Hansen GM, Brommage R. High-Throughput Screening of Mouse Gene Knockouts Identifies Established and Novel High Body Fat Phenotypes. Diabetes Metab Syndr Obes 2021; 14:3753-3785. [PMID: 34483672 PMCID: PMC8409770 DOI: 10.2147/dmso.s322083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/04/2021] [Indexed: 01/05/2023] Open
Abstract
PURPOSE Obesity is a major public health problem. Understanding which genes contribute to obesity may better predict individual risk and allow development of new therapies. Because obesity of a mouse gene knockout (KO) line predicts an association of the orthologous human gene with obesity, we reviewed data from the Lexicon Genome5000TM high throughput phenotypic screen (HTS) of mouse gene KOs to identify KO lines with high body fat. MATERIALS AND METHODS KO lines were generated using homologous recombination or gene trapping technologies. HTS body composition analyses were performed on adult wild-type and homozygous KO littermate mice from 3758 druggable mouse genes having a human ortholog. Body composition was measured by either DXA or QMR on chow-fed cohorts from all 3758 KO lines and was measured by QMR on independent high fat diet-fed cohorts from 2488 of these KO lines. Where possible, comparisons were made to HTS data from the International Mouse Phenotyping Consortium (IMPC). RESULTS Body fat data are presented for 75 KO lines. Of 46 KO lines where independent external published and/or IMPC KO lines are reported as obese, 43 had increased body fat. For the remaining 29 novel high body fat KO lines, Ksr2 and G2e3 are supported by data from additional independent KO cohorts, 6 (Asnsd1, Srpk2, Dpp8, Cxxc4, Tenm3 and Kiss1) are supported by data from additional internal cohorts, and the remaining 21 including Tle4, Ak5, Ntm, Tusc3, Ankk1, Mfap3l, Prok2 and Prokr2 were studied with HTS cohorts only. CONCLUSION These data support the finding of high body fat in 43 independent external published and/or IMPC KO lines. A novel obese phenotype was identified in 29 additional KO lines, with 27 still lacking the external confirmation now provided for Ksr2 and G2e3 KO mice. Undoubtedly, many mammalian obesity genes remain to be identified and characterized.
Collapse
Affiliation(s)
- David R Powell
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Jean-Pierre Revelli
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Deon D Doree
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Christopher M DaCosta
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Urvi Desai
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Melanie K Shadoan
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Lawrence Rodriguez
- Department of Information Technology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Michael Mullens
- Department of Information Technology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Qi M Yang
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Zhi-Ming Ding
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Laura L Kirkpatrick
- Department of Molecular Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Peter Vogel
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Brian Zambrowicz
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
- Department of Information Technology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
- Department of Molecular Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Arthur T Sands
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
- Department of Information Technology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
- Department of Molecular Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Kenneth A Platt
- Department of Molecular Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Gwenn M Hansen
- Department of Molecular Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Robert Brommage
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| |
Collapse
|
9
|
Reduced gonadotroph stimulation by ethanolamine plasmalogens in old bovine brains. Sci Rep 2021; 11:4757. [PMID: 33637828 PMCID: PMC7910589 DOI: 10.1038/s41598-021-84306-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 02/12/2021] [Indexed: 12/14/2022] Open
Abstract
Ethanolamine plasmalogens (EPls), unique alkenylacyl-glycerophospholipids, are the only known ligands of G-protein-coupled receptor 61—a novel receptor co-localised with gonadotropin-releasing hormone receptors on anterior pituitary gonadotrophs. Brain EPl decreases with age. Commercial EPl—extracted from the cattle brain (unidentified age)—can independently stimulate FSH secretion from gonadotrophs. We hypothesised that there exists an age-related difference in the quality, quantity, and ability of bovine brain EPls to stimulate bovine gonadotrophs. We compared the brains of young (about 26 month old heifers) and old (about 90 month old cows) Japanese Black bovines, including EPls obtained from both groups. Additionally, mRNA expressions of the EPl biosynthesis enzymes, glyceronephosphate O-acyltransferase, alkylglycerone phosphate synthase, and fatty acyl-CoA reductase 1 (FAR1) were evaluated in young and old hypothalami. The old-brain EPl did not stimulate FSH secretion from gonadotrophs, unlike the young-brain EPl. Molecular species of EPl were compared using two-dimensional liquid chromatography-mass spectrometry. We identified 20 EPl molecular species of which three and three exhibited lower (P < 0.05) and higher (P < 0.05) ratios, respectively, in old compared to young brains. In addition, quantitative reverse transcription-polymerase chain reaction detected higher FAR1 levels in the POA, but not in the ARC&ME tissues, of old cows than that of fertile young heifers. Therefore, old-brain EPl may be associated with age-related infertility.
Collapse
|
10
|
Watkins LR, Orlandi C. Orphan G Protein Coupled Receptors in Affective Disorders. Genes (Basel) 2020; 11:E694. [PMID: 32599826 PMCID: PMC7349732 DOI: 10.3390/genes11060694] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/20/2020] [Accepted: 06/21/2020] [Indexed: 12/12/2022] Open
Abstract
G protein coupled receptors (GPCRs) are the main mediators of signal transduction in the central nervous system. Therefore, it is not surprising that many GPCRs have long been investigated for their role in the development of anxiety and mood disorders, as well as in the mechanism of action of antidepressant therapies. Importantly, the endogenous ligands for a large group of GPCRs have not yet been identified and are therefore known as orphan GPCRs (oGPCRs). Nonetheless, growing evidence from animal studies, together with genome wide association studies (GWAS) and post-mortem transcriptomic analysis in patients, pointed at many oGPCRs as potential pharmacological targets. Among these discoveries, we summarize in this review how emotional behaviors are modulated by the following oGPCRs: ADGRB2 (BAI2), ADGRG1 (GPR56), GPR3, GPR26, GPR37, GPR50, GPR52, GPR61, GPR62, GPR88, GPR135, GPR158, and GPRC5B.
Collapse
Affiliation(s)
| | - Cesare Orlandi
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA;
| |
Collapse
|
11
|
Kadokawa H. Discovery of new receptors regulating luteinizing hormone and follicle-stimulating hormone secretion by bovine gonadotrophs to explore a new paradigm for mechanisms regulating reproduction. J Reprod Dev 2020; 66:291-297. [PMID: 32249236 PMCID: PMC7470908 DOI: 10.1262/jrd.2020-012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Previous studies in the 1960s and 1970s have reported that both gonadotropin-releasing hormone (GnRH) and estradiol-activated nuclear estrogen receptors regulate gonadotropin
secretion in women. However, I had previously reported that gonadotroph function is regulated by complex crosstalk between several membrane receptors. RNA-seq had previously
revealed 259 different receptor genes expressed in the anterior pituitary of heifers. However, the biological roles of most of these receptors remain unknown. I identified four new
receptors of interest: G protein-coupled receptor 30 (GPR30), anti-Mullerian hormone (AMH) receptor type 2 (AMHR2), and G protein-coupled receptors 61 and 153 (GPR61 and GPR153).
GPR30 rapidly (within a few minutes) mediates picomolar, but not nanomolar, levels of estradiol to suppress GnRH-induced luteinizing hormone (LH) secretion from bovine
gonadotrophs, without decreasing mRNA expressions of the LHα, LHβ, or follicle-stimulating hormone (FSH) β subunits. GPR30 is activated by other endogenous estrogens, estrone and
estriol. Moreover, GPR30 activation by zearalenone, a nonsteroidal mycoestrogen, suppresses LH secretion. AMHR2, activated by AMH, stimulates LH and FSH secretion, thus regulating
gonadotrophs, where other TGF-β family members, including inhibin and activin, potentially affect FSH secretion. I also show that GPR61, activated by its ligand (recently
discovered) significantly alters LH and FSH secretion. GPR61, GPR153, and AMHR2 co-localize with the GnRH receptor in unevenly dispersed areas of the bovine gonadotroph cell
surface, probably lipid rafts. The findings summarized in this review reveal a new paradigm regarding the mechanisms regulating reproduction via novel receptors expressed on bovine
gonadotrophs.
Collapse
Affiliation(s)
- Hiroya Kadokawa
- Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8515, Japan
| |
Collapse
|
12
|
Kereilwe O, Pandey K, Kadokawa H. Influence of brain plasmalogen changes on gonadotropin secretion from the cultured bovine anterior pituitary cells. Domest Anim Endocrinol 2018; 64:77-83. [PMID: 29754010 DOI: 10.1016/j.domaniend.2018.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 04/02/2018] [Accepted: 04/02/2018] [Indexed: 11/29/2022]
Abstract
We recently discovered that the orphan G-protein-coupled receptor (GPR) 61 colocalized with GnRH receptors (GnRHRs) on the surface of most of bovine gonadotrophs. A recent study suggested that ethanolamine plasmalogen (PI) is a ligand for GPR61 in mouse neuroblastoma. Therefore, this study evaluated the hypothesis that PI alters LH and FSH secretion from cultured bovine anterior pituitary (AP) cells. We prepared bovine AP cells from postpubertal heifers (26 mo old) and cultured the cells for 3.5 d. We treated the cells with increasing concentrations (0, 5, 50, 500, 5,000, 50,000, or 500,000 pg/mL) of phosphoethanolamine PI (PEPI) extracted from the bovine brain, or l-α-lysophosphatidylethanolamine PI (LEPI) extracted from the bovine brain, for 5 min before either no treatment or GnRH stimulation. The medium samples were harvested 2 h after culture for LH and FSH assays. Phosphoethanolamine PI (50-500 pg/mL) stimulated (P < 0.05) the basal secretion of FSH but not LH. Phosphoethanolamine PI at 50 pg/mL also enhanced (P < 0.05) GnRH-induced FSH secretion. However, higher doses (500-500,000 pg/mL) of PEPI suppressed GnRH-induced FSH secretion. Moreover, 50 to 500,000 pg/mL PEPI suppressed GnRH-induced LH secretion. None of the tested concentrations of LEPI showed any effect on basal or GnRH-induced LH or FSH secretion. Pretreatment with Sma and Mad pathway inhibitors suppressed FSH secretion induced by PEPI, whereas an extracellular signal-regulated kinase pathway inhibitor blocked the PEPI-induced suppression of GnRH-stimulated LH secretion. Therefore, PEPI, but not LEPI, extracted from the bovine brain, alters FSH and LH secretion from cultured AP cells. Further studies are required to decide whether PEPI binds to GPR61 and whether PEPI plays an important role in the control of gonadotropin secretion from gonadotrophs.
Collapse
Affiliation(s)
- O Kereilwe
- Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi-shi, Yamaguchi-ken, 1677-1, Japan
| | - K Pandey
- Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi-shi, Yamaguchi-ken, 1677-1, Japan
| | - H Kadokawa
- Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi-shi, Yamaguchi-ken, 1677-1, Japan.
| |
Collapse
|
13
|
Willmer T, Johnson R, Louw J, Pheiffer C. Blood-Based DNA Methylation Biomarkers for Type 2 Diabetes: Potential for Clinical Applications. Front Endocrinol (Lausanne) 2018; 9:744. [PMID: 30564199 PMCID: PMC6288427 DOI: 10.3389/fendo.2018.00744] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/23/2018] [Indexed: 12/22/2022] Open
Abstract
Type 2 diabetes (T2D) is a leading cause of death and disability worldwide. It is a chronic metabolic disorder that develops due to an interplay of genetic, lifestyle, and environmental factors. The biological onset of the disease occurs long before clinical symptoms develop, thus the search for early diagnostic and prognostic biomarkers, which could facilitate intervention strategies to prevent or delay disease progression, has increased considerably in recent years. Epigenetic modifications represent important links between genetic, environmental and lifestyle cues and increasing evidence implicate altered epigenetic marks such as DNA methylation, the most characterized and widely studied epigenetic mechanism, in the pathogenesis of T2D. This review provides an update of the current status of DNA methylation as a biomarker for T2D. Four databases, Scopus, Pubmed, Cochrane Central, and Google Scholar were searched for studies investigating DNA methylation in blood. Thirty-seven studies were identified, and are summarized with respect to population characteristics, biological source, and method of DNA methylation quantification (global, candidate gene or genome-wide). We highlight that differential methylation of the TCF7L2, KCNQ1, ABCG1, TXNIP, PHOSPHO1, SREBF1, SLC30A8, and FTO genes in blood are reproducibly associated with T2D in different population groups. These genes should be prioritized and replicated in longitudinal studies across more populations in future studies. Finally, we discuss the limitations faced by DNA methylation studies, which include including interpatient variability, cellular heterogeneity, and lack of accounting for study confounders. These limitations and challenges must be overcome before the implementation of blood-based DNA methylation biomarkers into a clinical setting. We emphasize the need for longitudinal prospective studies to support the robustness of the current findings of this review.
Collapse
Affiliation(s)
- Tarryn Willmer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- *Correspondence: Tarryn Willmer
| | - Rabia Johnson
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Johan Louw
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, Kwa-Dlangezwa, South Africa
| | - Carmen Pheiffer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
14
|
Kozielewicz P, Alomar H, Yusof S, Grafton G, Cooper AJ, Curnow SJ, Ironside JW, Pall H, Barnes NM. N-glycosylation and expression in human tissues of the orphan GPR61 receptor. FEBS Open Bio 2017; 7:1982-1993. [PMID: 29226084 PMCID: PMC5715243 DOI: 10.1002/2211-5463.12339] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/19/2017] [Accepted: 10/23/2017] [Indexed: 12/21/2022] Open
Abstract
A number of members of the G protein-coupled receptor class of cell surface receptors are 'orphans' with no known endogenous ligand. One of these orphan receptors is GPR61; there are little data about its expression in human cells and tissues. In this study, we investigated the post-translational modification of GPR61 by N-glycosylation at an identified consensus N-glycosylation site (N12) and the impact of this modification upon the subcellular expression of the protein. The N-glycosylation inhibitor tunicamycin reduced the apparent molecular weight of immunoreactivity associated with myc-tagged GPR61 by 1-2 kDa, which was comparable to the evident molecular weight of the myc-tagged N12S GPR61 mutant with disrupted consensus N-glycosylation site. Analysis of GPR61 expression demonstrated that tunicamycin treatment reduced considerably heterologous expression of GPR61 in the cell membrane despite the N12S GPR61 mutant being readily expressed at the cell surface. These results demonstrate that GPR61 is subject to N-glycosylation but suggest this is not a prerequisite for cell surface expression, although N-glycosylation of other proteins may be important for cell membrane expression of GPR61. Expression of GPR61 protein was demonstrated at the cellular level in human hippocampus and human peripheral blood mononuclear cells. In the latter, there was a significantly higher expression of GPR61 in the Th17 cell subset in comparison with resting CD4+ cells, which may point toward a potential role for the GPR61 receptor in autoimmune diseases. This is the first report that GPR61 protein is subject to post-translational modification and is expressed in immune cell subsets and the hippocampus. These findings will help guide studies to investigate the function of GPR61.
Collapse
Affiliation(s)
- Paweł Kozielewicz
- Institute of Clinical Sciences College of Medical and Dental Sciences University of Birmingham UK.,Present address: Department of Physiology and Pharmacology Karolinska Institutet Nanna Svartz väg 217 177 Stockholm Sweden
| | - Hatun Alomar
- Institute of Clinical Sciences College of Medical and Dental Sciences University of Birmingham UK.,Present address: Pharmacology and Toxicology Department College of Pharmacy King Saud University Riyadh 12372 Saudi Arabia
| | - Syaratul Yusof
- Institute of Clinical Sciences College of Medical and Dental Sciences University of Birmingham UK.,Present address: Faculty of Pharmacy Universiti Kebangsaan Malaysia 50300 Kuala Lumpur Malaysia
| | - Gillian Grafton
- Institute of Clinical Sciences College of Medical and Dental Sciences University of Birmingham UK
| | - Alison J Cooper
- Institute of Clinical Sciences College of Medical and Dental Sciences University of Birmingham UK
| | - S John Curnow
- Institute of Inflammation and Ageing College of Medical and Dental Sciences University of Birmingham UK
| | - James W Ironside
- National CJD Research and Surveillance Unit Centre for Clinical Brain Sciences University of Edinburgh UK
| | - Hardev Pall
- Neurology Department Queen Elizabeth Hospital Birmingham UK
| | - Nicholas M Barnes
- Institute of Clinical Sciences College of Medical and Dental Sciences University of Birmingham UK
| |
Collapse
|
15
|
Muroi T, Matsushima Y, Kanamori R, Inoue H, Fujii W, Yogo K. GPR62 constitutively activates cAMP signaling but is dispensable for male fertility in mice. Reproduction 2017; 154:755-764. [PMID: 28912303 DOI: 10.1530/rep-17-0333] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/05/2017] [Accepted: 09/14/2017] [Indexed: 11/08/2022]
Abstract
G-protein-coupled receptors (GPCRs) participate in diverse physiological functions and are promising targets for drug discovery. However, there are still over 140 orphan GPCRs whose functions remain to be elucidated. Gpr62 is one of the orphan GPCRs that is expressed in the rat and human brain. In this study, we found that Gpr62 is also expressed in male germ cells in mice, and its expression increases along with sperm differentiation. GPR62 lacks the BBXXB and DRY motifs, which are conserved across many GPCRs, and it was able to induce cAMP signaling in the absence of a ligand. These structural and functional features are conserved among mammals, and the mutant analysis of GPR62 has revealed that lacking of these motifs is involved in the constitutive activity. We also found that GPR62 can homooligomerize, but it is not sufficient for its constitutive activity. We further investigated its physiological function by using Gpr62 knockout (Gpr62-/-) mice. Gpr62-/- mice were born normally and did not show any abnormality in growth and behavior. In addition, both male and female Gp62-/- mice were fertile, and the differentiation and motility of spermatozoa were normal. We also found that Gpr61, the gene most similar to Gpr62 in the GPCR family shows a constitutive activity and an expression pattern similar to those of Gpr62 Our results suggest that GPR62 constitutively activates the cAMP pathway in male germ cells but is dispensable for male fertility, which is probably due to its functional redundancy with GPR61.
Collapse
Affiliation(s)
- Tomoyuki Muroi
- Department of AgricultureGraduate School of Integrated Science and Technology, Shizuoka University, Shizuoka, Japan
| | - Yuri Matsushima
- Department of Applied Biological ChemistryFaculty of Agriculture, Shizuoka University, Shizuoka, Japan
| | - Ryota Kanamori
- Department of Applied Biological ChemistryFaculty of Agriculture, Shizuoka University, Shizuoka, Japan
| | - Hikari Inoue
- Department of Applied Biological ChemistryFaculty of Agriculture, Shizuoka University, Shizuoka, Japan
| | - Wataru Fujii
- Department of Animal Resource SciencesGraduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Keiichiro Yogo
- Department of AgricultureGraduate School of Integrated Science and Technology, Shizuoka University, Shizuoka, Japan .,Department of Applied Biological ChemistryFaculty of Agriculture, Shizuoka University, Shizuoka, Japan.,College of AgricultureAcademic Institute, Shizuoka University, Shizuoka, Japan
| |
Collapse
|
16
|
Oishi A, Karamitri A, Gerbier R, Lahuna O, Ahmad R, Jockers R. Orphan GPR61, GPR62 and GPR135 receptors and the melatonin MT 2 receptor reciprocally modulate their signaling functions. Sci Rep 2017; 7:8990. [PMID: 28827538 PMCID: PMC5566548 DOI: 10.1038/s41598-017-08996-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 07/14/2017] [Indexed: 01/14/2023] Open
Abstract
Understanding the function of orphan G protein-coupled receptors (GPCRs), whose cognate ligand is unknown, is of major importance as GPCRs are privileged drug targets for many diseases. Recent phylogenetic studies classified three orphan receptors, GPR61, GPR62 and GPR135 among the melatonin receptor subfamily, but their capacity to bind melatonin and their biochemical functions are not well characterized yet. We show here that GPR61, GPR62 and GPR135 do not bind [3H]-melatonin nor 2-[125I]iodomelatonin and do not respond to melatonin in several signaling assays. In contrast, the three receptors show extensive spontaneous ligand-independent activities on the cAMP, inositol phosphate and ß-arrestin pathways with distinct pathway-specific profiles. Spontaneous ß-arrestin recruitment internalizes all three GPRs in the endosomal compartment. Co-expression of the melatonin binding MT2 receptor with GPR61, GPR62 or GPR135 has several consequences such as (i) the formation of receptor heteromers, (ii) the inhibition of melatonin-induced ß-arrestin2 recruitment to MT2 and (iii) the decrease of elevated cAMP levels upon melatonin stimulation in cells expressing spontaneously active GPR61 and GPR62. Collectively, these data show that GPR61, GPR62 and GPR135 are unable to bind melatonin, but show a reciprocal regulatory interaction with MT2 receptors.
Collapse
Affiliation(s)
- Atsuro Oishi
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS UMR, 8104, Paris, France.,University Paris Descartes, Paris, France
| | - Angeliki Karamitri
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS UMR, 8104, Paris, France.,University Paris Descartes, Paris, France
| | - Romain Gerbier
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS UMR, 8104, Paris, France.,University Paris Descartes, Paris, France
| | - Olivier Lahuna
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS UMR, 8104, Paris, France.,University Paris Descartes, Paris, France
| | - Raise Ahmad
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS UMR, 8104, Paris, France.,University Paris Descartes, Paris, France
| | - Ralf Jockers
- Inserm, U1016, Institut Cochin, Paris, France. .,CNRS UMR, 8104, Paris, France. .,University Paris Descartes, Paris, France.
| |
Collapse
|
17
|
Pandey K, Kereilwe O, Borromeo V, Kadokawa H. Heifers express G-protein coupled receptor 61 in anterior pituitary gonadotrophs in stage-dependent manner. Anim Reprod Sci 2017; 181:93-102. [DOI: 10.1016/j.anireprosci.2017.03.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/21/2017] [Accepted: 03/30/2017] [Indexed: 10/19/2022]
|
18
|
Khan MZ, He L. Neuro-psychopharmacological perspective of Orphan receptors of Rhodopsin (class A) family of G protein-coupled receptors. Psychopharmacology (Berl) 2017; 234:1181-1207. [PMID: 28289782 DOI: 10.1007/s00213-017-4586-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 02/27/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND In the central nervous system (CNS), G protein-coupled receptors (GPCRs) are the most fruitful targets for neuropsychopharmacological drug development. Rhodopsin (class A) is the most studied class of GPCR and includes orphan receptors for which the endogenous ligand is not known or is unclear. Characterization of orphan GPCRs has proven to be challenging, and the production pace of GPCR-based drugs has been incredibly slow. OBJECTIVE Determination of the functions of these receptors may provide unexpected insight into physiological and neuropathological processes. Advances in various methods and techniques to investigate orphan receptors including in situ hybridization and knockdown/knockout (KD/KO) showed extensive expression of these receptors in the mammalian brain and unmasked their physiological and neuropathological roles. Due to these rapid progress and development, orphan GPCRs are rising as a new and promising class of drug targets for neurodegenerative diseases and psychiatric disorders. CONCLUSION This review presents a neuropsychopharmacological perspective of 26 orphan receptors of rhodopsin (class A) family, namely GPR3, GPR6, GPR12, GPR17, GPR26, GPR35, GPR39, GPR48, GPR49, GPR50, GPR52, GPR55, GPR61, GPR62, GPR63, GPR68, GPR75, GPR78, GPR83, GPR84, GPR85, GPR88, GPR153, GPR162, GPR171, and TAAR6. We discussed the expression of these receptors in mammalian brain and their physiological roles. Furthermore, we have briefly highlighted their roles in neurodegenerative diseases and psychiatric disorders including Alzheimer's disease, Parkinson's disease, neuroinflammation, inflammatory pain, bipolar and schizophrenic disorders, epilepsy, anxiety, and depression.
Collapse
Affiliation(s)
- Muhammad Zahid Khan
- Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, Jiangsu Province, 210009, China.
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, Jiangsu Province, 210009, China
| |
Collapse
|
19
|
Felix JF, Bradfield JP, Monnereau C, van der Valk RJP, Stergiakouli E, Chesi A, Gaillard R, Feenstra B, Thiering E, Kreiner-Møller E, Mahajan A, Pitkänen N, Joro R, Cavadino A, Huikari V, Franks S, Groen-Blokhuis MM, Cousminer DL, Marsh JA, Lehtimäki T, Curtin JA, Vioque J, Ahluwalia TS, Myhre R, Price TS, Vilor-Tejedor N, Yengo L, Grarup N, Ntalla I, Ang W, Atalay M, Bisgaard H, Blakemore AI, Bonnefond A, Carstensen L, Eriksson J, Flexeder C, Franke L, Geller F, Geserick M, Hartikainen AL, Haworth CMA, Hirschhorn JN, Hofman A, Holm JC, Horikoshi M, Hottenga JJ, Huang J, Kadarmideen HN, Kähönen M, Kiess W, Lakka HM, Lakka TA, Lewin AM, Liang L, Lyytikäinen LP, Ma B, Magnus P, McCormack SE, McMahon G, Mentch FD, Middeldorp CM, Murray CS, Pahkala K, Pers TH, Pfäffle R, Postma DS, Power C, Simpson A, Sengpiel V, Tiesler CMT, Torrent M, Uitterlinden AG, van Meurs JB, Vinding R, Waage J, Wardle J, Zeggini E, Zemel BS, Dedoussis GV, Pedersen O, Froguel P, Sunyer J, Plomin R, Jacobsson B, Hansen T, Gonzalez JR, Custovic A, Raitakari OT, Pennell CE, Widén E, Boomsma DI, Koppelman GH, Sebert S, Järvelin MR, Hyppönen E, McCarthy MI, Lindi V, Harri N, Körner A, Bønnelykke K, Heinrich J, Melbye M, Rivadeneira F, Hakonarson H, Ring SM, Smith GD, Sørensen TIA, Timpson NJ, Grant SFA, Jaddoe VWV. Genome-wide association analysis identifies three new susceptibility loci for childhood body mass index. Hum Mol Genet 2016; 25:389-403. [PMID: 26604143 PMCID: PMC4854022 DOI: 10.1093/hmg/ddv472] [Citation(s) in RCA: 228] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 11/15/2015] [Indexed: 12/24/2022] Open
Abstract
A large number of genetic loci are associated with adult body mass index. However, the genetics of childhood body mass index are largely unknown. We performed a meta-analysis of genome-wide association studies of childhood body mass index, using sex- and age-adjusted standard deviation scores. We included 35 668 children from 20 studies in the discovery phase and 11 873 children from 13 studies in the replication phase. In total, 15 loci reached genome-wide significance (P-value < 5 × 10(-8)) in the joint discovery and replication analysis, of which 12 are previously identified loci in or close to ADCY3, GNPDA2, TMEM18, SEC16B, FAIM2, FTO, TFAP2B, TNNI3K, MC4R, GPR61, LMX1B and OLFM4 associated with adult body mass index or childhood obesity. We identified three novel loci: rs13253111 near ELP3, rs8092503 near RAB27B and rs13387838 near ADAM23. Per additional risk allele, body mass index increased 0.04 Standard Deviation Score (SDS) [Standard Error (SE) 0.007], 0.05 SDS (SE 0.008) and 0.14 SDS (SE 0.025), for rs13253111, rs8092503 and rs13387838, respectively. A genetic risk score combining all 15 SNPs showed that each additional average risk allele was associated with a 0.073 SDS (SE 0.011, P-value = 3.12 × 10(-10)) increase in childhood body mass index in a population of 1955 children. This risk score explained 2% of the variance in childhood body mass index. This study highlights the shared genetic background between childhood and adult body mass index and adds three novel loci. These loci likely represent age-related differences in strength of the associations with body mass index.
Collapse
Affiliation(s)
- Janine F Felix
- The Generation R Study Group, Department of Pediatrics, Department of Epidemiology,
| | | | - Claire Monnereau
- The Generation R Study Group, Department of Pediatrics, Department of Epidemiology
| | | | | | | | - Romy Gaillard
- The Generation R Study Group, Department of Pediatrics, Department of Epidemiology
| | - Bjarke Feenstra
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Elisabeth Thiering
- Institute of Epidemiology I, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany, Division of Metabolic and Nutritional Medicine, Dr von Hauner Children's Hospital, University of Munich Medical Center, Munich, Germany
| | - Eskil Kreiner-Møller
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital
| | | | - Niina Pitkänen
- Research Centre of Applied and Preventive Cardiovascular Medicine, Institute of Clinical Medicine, Neurology
| | - Raimo Joro
- Institute of Biomedicine, Physiology, University of Eastern Finland, Kuopio, Finland
| | - Alana Cavadino
- Centre for Environmental and Preventive Medicine, Wolfson Institute of Preventive Medicine, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK, Population, Policy and Practice, UCL Institute of Child Health
| | | | - Steve Franks
- Institute of Reproductive and Developmental Biology
| | - Maria M Groen-Blokhuis
- Department of Biological Psychology, VU University Amsterdam, NCA Neuroscience Campus Amsterdam, EMGO+ Institute for Health and Care Research, Amsterdam, the Netherlands
| | - Diana L Cousminer
- Institute for Molecular Medicine, Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Julie A Marsh
- School of Women's and Infants' Health, The University of Western Australia, Perth, Australia
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland, Department of Clinical Chemistry
| | - John A Curtin
- Centre for Respiratory Medicine and Allergy, Institute of Inflammation and Repair, University of Manchester and University Hospital of South Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Jesus Vioque
- Universidad Miguel Hernandez, Elche-Alicante, Spain, CIBER Epidemiología y Salud Pública (CIBERESP), Spain
| | - Tarunveer S Ahluwalia
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark, Steno Diabetes Center, Gentofte, Denmark
| | - Ronny Myhre
- Department of Genes and Envrionment, Division of Epidemiology
| | - Thomas S Price
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, USA
| | - Natalia Vilor-Tejedor
- CIBER Epidemiología y Salud Pública (CIBERESP), Spain, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain, Pompeu Fabra University (UPF), Barcelona, Spain
| | - Loïc Yengo
- CNRS UMR8199, Pasteur Institute Lille, France, European Genomic Institute for Diabetes (EGID), Lille, France
| | - Niels Grarup
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ioanna Ntalla
- Department of Health Sciences, University of Leicester, Leicester, UK, Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Wei Ang
- School of Women's and Infants' Health, The University of Western Australia, Perth, Australia
| | - Mustafa Atalay
- Institute of Biomedicine, Physiology, University of Eastern Finland, Kuopio, Finland
| | - Hans Bisgaard
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital
| | - Alexandra I Blakemore
- Section of Investigative Medicine, Division of Diabetes, Endocrinology, and Metabolism, Faculty of Medicine, Imperial College, London, UK
| | - Amelie Bonnefond
- CNRS UMR8199, Pasteur Institute Lille, France, European Genomic Institute for Diabetes (EGID), Lille, France
| | - Lisbeth Carstensen
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | | | | | - Johan Eriksson
- National Institute for Health and Welfare, Helsinki, Finland
| | - Claudia Flexeder
- Institute of Epidemiology I, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Frank Geller
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Mandy Geserick
- Center of Pediatric Research, Department of Women's and Child Health, LIFE Child (Leipzig Research Center for Civilization Diseases)
| | | | | | - Joel N Hirschhorn
- Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, USA, Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, USA, Department of Genetics, Harvard Medical School, Boston, USA
| | - Albert Hofman
- The Generation R Study Group, Department of Epidemiology
| | - Jens-Christian Holm
- The Children's Obesity Clinic, Department of Pediatrics, Copenhagen University Hospital Holbæk, The Danish Childhood Obesity Biobank, Denmark, Institute of Medicine, Copenhagen University, Copenhagen, Denmark
| | - Momoko Horikoshi
- Wellcome Trust Centre for Human Genetics, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Jouke Jan Hottenga
- Department of Biological Psychology, VU University Amsterdam, NCA Neuroscience Campus Amsterdam, EMGO+ Institute for Health and Care Research, Amsterdam, the Netherlands
| | - Jinyan Huang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haja N Kadarmideen
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Mika Kähönen
- Department of Clinical Physiology, University of Tampere School of Medicine, Tampere, Finland, Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
| | - Wieland Kiess
- Center of Pediatric Research, Department of Women's and Child Health
| | - Hanna-Maaria Lakka
- Institute of Biomedicine, Physiology, University of Eastern Finland, Kuopio, Finland
| | - Timo A Lakka
- Institute of Biomedicine, Physiology, University of Eastern Finland, Kuopio, Finland, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland, Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Alexandra M Lewin
- Department of Epidemiology and Biostatistics, MRC Health Protection Agency (HPE) Centre for Environment and Health, School of Public Health, Imperial College London, UK
| | - Liming Liang
- Department of Epidemiology, Department of Biostatistics, Harvard School of Public Health, Boston, USA
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland, Department of Clinical Chemistry
| | - Baoshan Ma
- College of Information Science and Technology, Dalian Maritime University, Dalian, Liaoning Province, China
| | - Per Magnus
- Division of Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | - Shana E McCormack
- Division of Human Genetics, Division of Endocrinology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - George McMahon
- MRC Integrative Epidemiology Unit at the University of Bristol
| | | | - Christel M Middeldorp
- Department of Biological Psychology, VU University Amsterdam, NCA Neuroscience Campus Amsterdam, EMGO+ Institute for Health and Care Research, Amsterdam, the Netherlands
| | - Clare S Murray
- Centre for Respiratory Medicine and Allergy, Institute of Inflammation and Repair, University of Manchester and University Hospital of South Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Katja Pahkala
- Research Centre of Applied and Preventive Cardiovascular Medicine, Department of Health and Physical Activity, Paavo Nurmi Centre, Sports and Exercise Medicine Unit
| | - Tune H Pers
- Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, USA, Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, USA
| | - Roland Pfäffle
- Center of Pediatric Research, Department of Women's and Child Health, CrescNet, Medical Faculty, University of Leipzig, Germany
| | - Dirkje S Postma
- Department of Pulmonology, GRIAC (Groningen Research Institute for Asthma and COPD)
| | - Christine Power
- Population, Policy and Practice, UCL Institute of Child Health
| | - Angela Simpson
- Centre for Respiratory Medicine and Allergy, Institute of Inflammation and Repair, University of Manchester and
| | - Verena Sengpiel
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, Sahlgrenska University Hosptial, Gothenburg, Sweden
| | - Carla M T Tiesler
- Institute of Epidemiology I, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany, Division of Metabolic and Nutritional Medicine, Dr von Hauner Children's Hospital, University of Munich Medical Center, Munich, Germany
| | - Maties Torrent
- CIBER Epidemiología y Salud Pública (CIBERESP), Spain, Area de Salut de Menorca, ib-salut, Menorca, Spain
| | - André G Uitterlinden
- The Generation R Study Group, Department of Epidemiology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Joyce B van Meurs
- Department of Epidemiology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rebecca Vinding
- Department of Pediatrics, Naestved Hospital, Naestved, Denmark, COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital
| | - Johannes Waage
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital
| | - Jane Wardle
- Department of Epidemiology and Public Health, University College London, UK
| | - Eleftheria Zeggini
- Wellcome Trust Sanger Institute, The Morgan Building, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, UK
| | - Babette S Zemel
- Division of Gastroenterology, Hepatology and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA, USA, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - George V Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Oluf Pedersen
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Philippe Froguel
- CNRS UMR8199, Pasteur Institute Lille, France, Department of Genomics of Common Disease, School of Public Health, Imperial College London, Hammersmith Hospital, London, UK
| | - Jordi Sunyer
- CIBER Epidemiología y Salud Pública (CIBERESP), Spain, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain, Pompeu Fabra University (UPF), Barcelona, Spain, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Robert Plomin
- King's College London, MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, De Crespigny Park, London, UK
| | - Bo Jacobsson
- Department of Genes and Envrionment, Division of Epidemiology, Department of Obstetrics and Gynecology, Sahlgrenska Academy, Sahlgrenska University Hosptial, Gothenburg, Sweden
| | - Torben Hansen
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Juan R Gonzalez
- CIBER Epidemiología y Salud Pública (CIBERESP), Spain, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain, Pompeu Fabra University (UPF), Barcelona, Spain
| | - Adnan Custovic
- Centre for Respiratory Medicine and Allergy, Institute of Inflammation and Repair, University of Manchester and University Hospital of South Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Olli T Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, Department of Clinical Physiology and Nuclear Medicine
| | - Craig E Pennell
- School of Women's and Infants' Health, The University of Western Australia, Perth, Australia
| | - Elisabeth Widén
- Institute for Molecular Medicine, Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Dorret I Boomsma
- Department of Biological Psychology, VU University Amsterdam, NCA Neuroscience Campus Amsterdam, EMGO+ Institute for Health and Care Research, Amsterdam, the Netherlands
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, GRIAC (Groningen Research Institute for Asthma and COPD), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sylvain Sebert
- Centre for Life Course Epidemiology, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Marjo-Riitta Järvelin
- Centre for Life Course Epidemiology, Biocenter Oulu, University of Oulu, Oulu, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection Agency (HPE) Centre for Environment and Health, School of Public Health, Imperial College London, UK, Unit of Primary Care, Oulu University Hospital, Oulu, Finland, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland
| | - Elina Hyppönen
- Population, Policy and Practice, UCL Institute of Child Health, School of Population Health and Sansom Institute, University of South Australia, Adelaide, Australia, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Mark I McCarthy
- Wellcome Trust Centre for Human Genetics, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK, Oxford National Institute for Health Research (NIHR) Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Virpi Lindi
- Institute of Biomedicine, Physiology, University of Eastern Finland, Kuopio, Finland
| | - Niinikoski Harri
- Department of Pediatrics, Turku University Hospital, University of Turku, Turku, Finland
| | - Antje Körner
- Center of Pediatric Research, Department of Women's and Child Health
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital
| | - Joachim Heinrich
- Institute of Epidemiology I, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Mads Melbye
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA and
| | - Fernando Rivadeneira
- The Generation R Study Group, Department of Epidemiology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Hakon Hakonarson
- Center for Applied Genomics, Division of Human Genetics, Department of Obstetrics and Gynecology, Sahlgrenska Academy, Sahlgrenska University Hosptial, Gothenburg, Sweden
| | - Susan M Ring
- MRC Integrative Epidemiology Unit at the University of Bristol, Avon Longitudinal Study of Parents and Children (ALSPAC), School of Social and Community Medicine, University of Bristol, Bristol, UK
| | | | - Thorkild I A Sørensen
- MRC Integrative Epidemiology Unit at the University of Bristol, Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark, Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospital, The Capital Region, Copenhagen, Denmark
| | | | - Struan F A Grant
- Center for Applied Genomics, Division of Human Genetics, Division of Endocrinology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vincent W V Jaddoe
- The Generation R Study Group, Department of Pediatrics, Department of Epidemiology
| | | | | |
Collapse
|
20
|
Yuan W, Xia Y, Bell CG, Yet I, Ferreira T, Ward KJ, Gao F, Loomis AK, Hyde CL, Wu H, Lu H, Liu Y, Small KS, Viñuela A, Morris AP, Berdasco M, Esteller M, Brosnan MJ, Deloukas P, McCarthy MI, John SL, Bell JT, Wang J, Spector TD. An integrated epigenomic analysis for type 2 diabetes susceptibility loci in monozygotic twins. Nat Commun 2014; 5:5719. [PMID: 25502755 PMCID: PMC4284644 DOI: 10.1038/ncomms6719] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 10/31/2014] [Indexed: 01/05/2023] Open
Abstract
DNA methylation has a great potential for understanding the aetiology of common complex traits such as Type 2 diabetes (T2D). Here we perform genome-wide methylated DNA immunoprecipitation sequencing (MeDIP-seq) in whole-blood-derived DNA from 27 monozygotic twin pairs and follow up results with replication and integrated omics analyses. We identify predominately hypermethylated T2D-related differentially methylated regions (DMRs) and replicate the top signals in 42 unrelated T2D cases and 221 controls. The strongest signal is in the promoter of the MALT1 gene, involved in insulin and glycaemic pathways, and related to taurocholate levels in blood. Integrating the DNA methylome findings with T2D GWAS meta-analysis results reveals a strong enrichment for DMRs in T2D-susceptibility loci. We also detect signals specific to T2D-discordant twins in the GPR61 and PRKCB genes. These replicated T2D associations reflect both likely causal and consequential pathways of the disease. The analysis indicates how an integrated genomics and epigenomics approach, utilizing an MZ twin design, can provide pathogenic insights as well as potential drug targets and biomarkers for T2D and other complex traits. Type 2 diabetes (T2D) is a highly heterogeneous disease with a strong genetic component. Here the authors examine genome-wide methylation patterns in T2D-discordant, T2D-concordant and healthy concordant monozygotic twin pairs, and identify DNA methylation signals that may represent new biomarkers or drug targets for T2D.
Collapse
Affiliation(s)
- Wei Yuan
- Department of Twin Research &Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | | | - Christopher G Bell
- Department of Twin Research &Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Idil Yet
- Department of Twin Research &Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Teresa Ferreira
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Kirsten J Ward
- Department of Twin Research &Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Fei Gao
- BGI-Shenzhen, Shenzhen 518083, China
| | - A Katrina Loomis
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, USA
| | - Craig L Hyde
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, USA
| | | | - Hanlin Lu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Yuan Liu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Kerrin S Small
- Department of Twin Research &Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Ana Viñuela
- Department of Twin Research &Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Andrew P Morris
- 1] Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK [2] Department of Biostatistics, University of Liverpool, Liverpool L69 3GA, UK
| | - María Berdasco
- Cancer Epigenetics and Biology Program (PEBC), Hospital Duran i Reynals, Barcelona 08908, Spain
| | - Manel Esteller
- 1] Cancer Epigenetics and Biology Program (PEBC), Hospital Duran i Reynals, Barcelona 08908, Spain [2] Department of Physiological Sciences II, School of Medicine, University of Barcelona, Barcelona, Catalonia 08907, Spain [3] Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia 08010, Spain
| | - M Julia Brosnan
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, USA
| | - Panos Deloukas
- 1] William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK [2] King Abdulaziz University, Jeddah 22254, Saudi Arabia
| | - Mark I McCarthy
- 1] Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK [2] Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, oxford OX3 7LE, UK [3] Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford OX3 7LE, UK
| | - Sally L John
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, USA
| | - Jordana T Bell
- Department of Twin Research &Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Jun Wang
- 1] BGI-Shenzhen, Shenzhen 518083, China [2] King Abdulaziz University, Jeddah 22254, Saudi Arabia [3] Department of Biology, University of Copenhagen, Copenhagen DK-2200, Denmark [4] The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Tim D Spector
- Department of Twin Research &Genetic Epidemiology, King's College London, London SE1 7EH, UK
| |
Collapse
|
21
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: G protein-coupled receptors. Br J Pharmacol 2013; 170:1459-581. [PMID: 24517644 PMCID: PMC3892287 DOI: 10.1111/bph.12445] [Citation(s) in RCA: 505] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. G protein-coupled receptors are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, catalytic receptors, nuclear hormone receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|